Skip to main content

Advertisement

Log in

STEM CELL BIOLOGY

Molecular landscape of immune pressure and escape in aplastic anemia

  • Article
  • Published:
Leukemia Submit manuscript

Abstract

Idiopathic aplastic anemia (IAA) pathophysiology is dominated by autoreactivity of human leukocyte antigen (HLA)-restricted T-cells against antigens presented by hematopoietic stem and progenitor cells (HSPCs). Expansion of PIGA and HLA class I mutant HSPCs have been linked to immune evasion from T-cell mediated pressures. We hypothesized that in analogy with antitumor immunity, the pathophysiological cascade of immune escape in IAA is initiated by immunoediting pressures and culminates with mechanisms of clonal evolution characterized by hits in immune recognition and response genes. To that end, we studied the genetic and transcriptomic make-up of the antigen presentation complexes in a large cohort of patients with IAA and paroxysmal nocturnal hemoglobinuria (PNH) by using single-cell RNA, high throughput DNA sequencing and single nucleotide polymorphism (SNP)-array platforms. At disease onset, HSPCs displayed activation of selected HLA class I and II-restricted mechanisms, without extensive inhibition of immune checkpoint apparatus. Using a newly implemented bioinformatic framework we found that not only class I but also class II genes were often impaired by acquisition of genetic aberrations. We also demonstrated the presence of novel somatic alterations in immune genes possibly contributing to the evasion from the autoimmune T-cells. In contrast, these hits were absent in myeloid neoplasia. These aberrations were not mutually exclusive with PNH and did not correlate with the accumulation of myeloid-driver hits. Our findings shed light on the mechanisms of immune activation and escape in IAA and define alternative modes of clonal hematopoiesis.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Study design.
Fig. 2: Single-cell RNAseq analysis of antigen presentation machinery genes in HSPCs in aplastic anemia patients and healthy controls.
Fig. 3: Landscape of HLA aberrations.
Fig. 4: Immune, PNH and myeloid landscape.

Similar content being viewed by others

References

  1. Young NS. Current concepts in the pathophysiology and treatment of aplastic anemia. Hematology. 2013;2013:76–81.

    Article  Google Scholar 

  2. Risitano AM. Oligoclonal and polyclonal CD4 and CD8 lymphocytes in aplastic anemia and paroxysmal nocturnal hemoglobinuria measured by Vbeta CDR3 spectra typing and flow cytometry. Blood. 2002;100:178–83.

    Article  CAS  Google Scholar 

  3. Sloand E. Intracellular interferon-gamma in circulating and marrow T cells detected by flow cytometry and the response to immunosuppressive therapy in patients with aplastic anemia. Blood. 2002;100:1185–91.

    Article  CAS  Google Scholar 

  4. Kordasti S, Marsh J, Al-Khan S, Jiang J, Smith A, Mohamedali A, et al. Functional characterization of CD4+ T cells in aplastic anemia. Blood. 2012;119:2033–43.

    Article  CAS  Google Scholar 

  5. de Latour RP, Visconte V, Takaku T, Wu C, Erie AJ, Sarcon AK, et al. Th17 immune responses contribute to the pathophysiology of aplastic anemia. Blood. 2010;116:4175–84.

    Article  Google Scholar 

  6. Giudice V, Selleri C. Aplastic anemia: pathophysiology. Semin Hematol. 2022;59:13–20.

    Article  Google Scholar 

  7. Pagliuca S, Gurnari C, Awada H, Kishtagari A, Kongkiatkamon S, Terkawi L, et al. The similarity of class II HLA genotypes defines patterns of autoreactivity in idiopathic bone marrow failure disorders. Blood. 2021. https://doi.org/10.1182/blood.2021012900.

  8. Kook H, Risitano AM, Zeng W, Wlodarski M, Lottemann C, Nakamura R, et al. Changes in T-cell receptor VB repertoire in aplastic anemia: effects of different immunosuppressive regimens. Blood. 2002;99:3668–75.

    Article  CAS  Google Scholar 

  9. Giudice V, Feng X, Lin Z, Hu W, Zhang F, Qiao W, et al. Deep sequencing and flow cytometric characterization of expanded effector memory CD8+CD57+ T cells frequently reveals T-cell receptor Vβ oligoclonality and CDR3 homology in acquired aplastic anemia. Haematologica. 2018;103:759–69.

    Article  CAS  Google Scholar 

  10. Patel BA, Giudice V, Young NS. Immunologic effects on the haematopoietic stem cell in marrow failure. Best Pract Res Clin Haematol. 2021;34:101276.

    Article  CAS  Google Scholar 

  11. Yoshizato T, Dumitriu B, Hosokawa K, Makishima H, Yoshida K, Townsley D, et al. Somatic mutations and clonal hematopoiesis in aplastic anemia. N Engl J Med. 2015;373:35–47.

    Article  CAS  Google Scholar 

  12. Kulasekararaj AG, Jiang J, Smith AE, Mohamedali AM, Mian S, Gandhi S, et al. Somatic mutations identify a subgroup of aplastic anemia patients who progress to myelodysplastic syndrome. Blood. 2014;124:2698–704.

    Article  CAS  Google Scholar 

  13. Gurnari C, Pagliuca S, Prata PH, Galimard J-E, Catto LFB, Larcher L, et al. Clinical and molecular determinants of clonal evolution in aplastic anemia and paroxysmal nocturnal hemoglobinuria. J Clin Oncol. 2022. https://doi.org/10.1200/JCO.22.00710.

  14. Babushok DV. A brief, but comprehensive, guide to clonal evolution in aplastic anemia. Hematol Am Soc Hematol Educ Program. 2018;2018:457–66.

    Article  Google Scholar 

  15. Sun L, Babushok DV. Secondary myelodysplastic syndrome and leukemia in acquired aplastic anemia and paroxysmal nocturnal hemoglobinuria. Blood. 2020;136:36–49.

    Article  Google Scholar 

  16. Tiu R, Gondek L, O’Keefe C, Maciejewski JP. Clonality of the stem cell compartment during evolution of myelodysplastic syndromes and other bone marrow failure syndromes. Leukemia. 2007;21:1648–57.

    Article  CAS  Google Scholar 

  17. Young NS, Maciejewski JP. Genetic and environmental effects in paroxysmal nocturnal hemoglobinuria: this little PIG-A goes ‘Why? Why? Why?’. J Clin Investig. 2000;106:637–41.

    Article  CAS  Google Scholar 

  18. Babushok DV, Duke JL, Xie HM, Stanley N, Atienza J, Perdigones N, et al. Somatic HLA mutations expose the role of class I-mediated autoimmunity in aplastic anemia and its clonal complications. Blood Adv. 2017;1:1900–10.

    Article  CAS  Google Scholar 

  19. Katagiri T, Sato-Otsubo A, Kashiwase K, Morishima S, Sato Y, Mori Y, et al. Frequent loss of HLA alleles associated with copy number-neutral 6pLOH in acquired aplastic anemia. Blood. 2011;118:6601–9.

    Article  CAS  Google Scholar 

  20. Mizumaki H, Hosomichi K, Hosokawa K, Yoroidaka T, Imi T, Zaimoku Y, et al. A frequent nonsense mutation in exon 1 across certain HLA-A and -B alleles in leukocytes of patients with acquired aplastic anemia. Haematologica. 2020. https://doi.org/10.3324/haematol.2020.247809.

  21. Imi T, Katagiri T, Hosomichi K, Zaimoku Y, Hoang Nguyen V, Nakagawa N, et al. Sustained clonal hematopoiesis by HLA-lacking hematopoietic stem cells without driver mutations in aplastic anemia. Blood Adv. 2018;2:1000–12.

    Article  CAS  Google Scholar 

  22. McGranahan N, Rosenthal R, Hiley CT, Rowan AJ, Watkins TBK, Wilson GA, et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell. 2017;171:1259–71.e11.

    Article  CAS  Google Scholar 

  23. Christopher MJ, Petti AA, Rettig MP, Miller CA, Chendamarai E, Duncavage EJ, et al. Immune escape of relapsed AML cells after allogeneic transplantation. N Engl J Med. 2018;379:2330–41.

    Article  CAS  Google Scholar 

  24. Jiménez P, Cantón J, Collado A, Cabrera T, Serrano A, Real LM, et al. Chromosome loss is the most frequent mechanism contributing to HLA haplotype loss in human tumors. Int J Cancer. 1999;83:91–97.

    Article  Google Scholar 

  25. Vago L, Perna SK, Zanussi M, Mazzi B, Barlassina C, Stanghellini MTL, et al. Loss of mismatched HLA in leukemia after stem-cell transplantation. N Engl J Med. 2009;361:478–88.

    Article  CAS  Google Scholar 

  26. Afable MG, Wlodarski M, Makishima H, Shaik M, Sekeres MA, Tiu RV, et al. SNP array-based karyotyping: differences and similarities between aplastic anemia and hypocellular myelodysplastic syndromes. Blood. 2011;117:6876–84.

    Article  CAS  Google Scholar 

  27. Zaimoku Y, Takamatsu H, Hosomichi K, Ozawa T, Nakagawa N, Imi T, et al. Identification of an HLA class I allele closely involved in the autoantigen presentation in acquired aplastic anemia. Blood. 2017;129:2908–16.

    Article  CAS  Google Scholar 

  28. Seliger B, Maeurer MJ, Ferrone S. Antigen-processing machinery breakdown and tumor growth. Immunol Today. 2000;21:455–64.

    Article  CAS  Google Scholar 

  29. Toffalori C, Zito L, Gambacorta V, Riba M, Oliveira G, Bucci G, et al. Immune signature drives leukemia escape and relapse after hematopoietic cell transplantation. Nat Med. 2019;25:603–11.

    Article  CAS  Google Scholar 

  30. Tomasi TB, Magner WJ, Khan ANH. Epigenetic regulation of immune escape genes in cancer. Cancer Immunol Immunother. 2006;55:1159–84.

    Article  Google Scholar 

  31. Norde WJ, Maas F, Hobo W, Korman A, Quigley M, Kester MGD, et al. PD-1/PD-L1 interactions contribute to functional T-cell impairment in patients who relapse with cancer after allogeneic stem cell transplantation. Cancer Res. 2011;71:5111–22.

    Article  CAS  Google Scholar 

  32. O’Sullivan T, Saddawi-Konefka R, Vermi W, Koebel CM, Arthur C, White JM, et al. Cancer immunoediting by the innate immune system in the absence of adaptive immunity. J Exp Med. 2012;209:1869–82.

    Article  Google Scholar 

  33. Zhu C, Lian Y, Wang C, Wu P, Li X, Gao Y, et al. Single-cell transcriptomics dissects hematopoietic cell destruction and T-cell engagement in aplastic anemia. Blood. 2021;138:23–33.

    Article  CAS  Google Scholar 

  34. Alsaggaf R, Katta S, Wang T, Hicks BD, Zhu B, Spellman SR, et al. Epigenetic aging and hematopoietic cell transplantation in patients with severe aplastic anemia. Transpl Cell Ther. 2021;27:313.e1–313.e8.

    Article  CAS  Google Scholar 

  35. Savage SA, Viard M, O’hUigin C, Zhou W, Yeager M, Li SA, et al. Genome-wide association study identifies HLA-DPB1 as a significant risk factor for severe aplastic anemia. Am J Hum Genet. 2020;106:264–71.

    Article  CAS  Google Scholar 

  36. Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–76.

    Article  CAS  Google Scholar 

  37. Robinson J, Barker DJ, Georgiou X, Cooper MA, Flicek P, Marsh SGE. IPD-IMGT/HLA database. Nucleic Acids Res. 2019;gkz950.

  38. Li L, Dong J, Yan L, Yong J, Liu X, Hu Y, et al. Single-cell RNA-seq analysis maps development of human germline cells and gonadal niche interactions. Cell Stem Cell. 2017;20:858–73.e4.

    Article  CAS  Google Scholar 

  39. Gao S, Yan L, Wang R, Li J, Yong J, Zhou X, et al. Tracing the temporal-spatial transcriptome landscapes of the human fetal digestive tract using single-cell RNA-sequencing. Nat Cell Biol. 2018;20:721–34.

    Article  CAS  Google Scholar 

  40. Alessandrì L, Arigoni M, Calogero R. Differential expression analysis in single-cell transcriptomics. In: Proserpio V (ed). Single cell methods. Springer New York: New York, NY, 2019, pp 425–32.

  41. Fine JP, Gray RJ. A proportional hazards model for the subdistribution of a competing risk. J Am Stat Assoc. 1999;94:496–509.

    Article  Google Scholar 

  42. Pagliuca S, Gurnari C, Rubio MT, Visconte V, Lenz TL. Individual HLA heterogeneity and its implications for cellular immune evasion in cancer and beyond. Front Immunol. 2022;13:944872.

    Article  CAS  Google Scholar 

  43. Maciejewski JP, Follmann D, Nakamura R, Saunthararajah Y, Rivera CE, Simonis T, et al. Increased frequency of HLA-DR2 in patients with paroxysmal nocturnal hemoglobinuria and the PNH/aplastic anemia syndrome. Blood. 2001;98:3513–9.

    Article  CAS  Google Scholar 

  44. Zeng W, Nakao S, Takamatsu H, Yachie A, Takami A, Kondo Y, et al. Characterization of T-cell repertoire of the bone marrow in immune-mediated aplastic anemia: evidence for the involvement of antigen-driven T-cell response in cyclosporine-dependent aplastic anemia. Blood. 1999;93:3008–16.

    Article  CAS  Google Scholar 

  45. Nakao S, Takamatsu H, Yachie A, Itoh T, Yamaguchi M, Ueda M, et al. Establishment of a CD4+ T cell clone recognizing autologous hematopoietic progenitor cells from a patient with immune-mediated aplastic anemia. Exp Hematol. 1995;23:433–8.

    CAS  Google Scholar 

  46. Tsuji N, Hosokawa K, Urushihara R, Tanabe M, Takamatsu H, Ishiyama K, et al. Epigenetic loss of the HLA-DR15 expression on hematopoietic stem progenitor cells in patients with acquired aplastic anemia characterized by cyclosporine dependency: a novel mechanism underlying the immune escape of hematopoietic stem progenitor cells. Blood. 2020;136:23–24.

    Article  Google Scholar 

  47. He MY, Kridel R. Treatment resistance in diffuse large B-cell lymphoma. Leukemia. 2021;35:2151–65.

    Article  Google Scholar 

  48. Johansson P, Klein-Hitpass L, Röth A, Möllmann M, Reinhardt HC, Dührsen U, et al. Mutations in PIGA cause a CD52-/GPI-anchor-deficient phenotype complicating alemtuzumab treatment in T-cell prolymphocytic leukemia. Eur J Haematol. 2020;105:786–96.

    Article  CAS  Google Scholar 

  49. Loeff FC, Falkenburg JHF, Hageman L, Huisman W, Veld SAJ, van Egmond HME, et al. High mutation frequency of the PIGA gene in T cells results in reconstitution of GPI anchor /CD52 T cells that can give early immune protection after alemtuzumab-based T cell-depleted allogeneic stem. Cell Transplant JI. 2018;200:2199–208.

    CAS  Google Scholar 

  50. Jia D, Augert A, Kim D-W, Eastwood E, Wu N, Ibrahim AH, et al. Crebbp loss drives small cell lung cancer and increases sensitivity to HDAC inhibition. Cancer Discov. 2018;8:1422–37.

    Article  CAS  Google Scholar 

  51. Morin RD, Mendez-Lago M, Mungall AJ, Goya R, Mungall KL, Corbett RD, et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature. 2011;476:298–303.

    Article  CAS  Google Scholar 

  52. Robinson DR, Wu Y-M, Lonigro RJ, Vats P, Cobain E, Everett J, et al. Integrative clinical genomics of metastatic cancer. Nature. 2017;548:297–303.

    Article  CAS  Google Scholar 

  53. Hosokawa K, Mizumaki H, Yoroidaka T, Maruyama H, Imi T, Tsuji N, et al. HLA class I allele-lacking leukocytes predict rare clonal evolution to MDS/AML in patients with acquired aplastic anemia. Blood. 2021;137:3576–80.

    Article  CAS  Google Scholar 

  54. Zaimoku Y, Patel BA, Adams SD, Shalhoub R, Groarke EM, Lee AAC, et al. HLA associations, somatic loss of HLA expression, and clinical outcomes in immune aplastic anemia. Blood. 2021;138:2799–809.

    Article  CAS  Google Scholar 

  55. Groarke EM, Patel BA, Shalhoub R, Gutierrez-Rodrigues F, Desai P, Leuva H, et al. Predictors of clonal evolution and myeloid neoplasia following immunosuppressive therapy in severe aplastic anemia. Leukemia. 2022. https://doi.org/10.1038/s41375-022-01636-8.

  56. Montesion M, Murugesan K, Jin DX, Sharaf R, Sanchez N, Guria A, et al. Somatic HLA class I loss is a widespread mechanism of immune evasion which refines the use of tumor mutational burden as a biomarker of checkpoint inhibitor response. Cancer Discov. 2021;11:282–92.

    Article  CAS  Google Scholar 

  57. Shukla SA, Rooney MS, Rajasagi M, Tiao G, Dixon PM, Lawrence MS, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol. 2015;33:1152–8.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by US National Institute of Health (NIH) grants R35 HL135795, R01HL123904, R01 380HL118281, R01 HL128425, R01 HL132071, Edward P. Evans Foundation, The Leukemia & Lymphoma Society TRP Award 6645-22 (to JPM), Aplastic Anemia and MDS International Foundation, Italian Society of Hematology, Fondation ARC pour la Recherche sur le Cancer, Association HPN France – Aplasie medullaire and Foundation For Rare Diseases (FFRD) (to SP), VeloSano Pilot Award and Vera and Joseph Dresner Foundation–MDS (to VV), Edward P. Evans Foundation (to CG). The CIBMTR is supported primarily by Public Health Service U24CA076518 from the National Cancer Institute (NCI), the National Heart, Lung and Blood Institute (NHLBI) and the National Institute of Allergy and Infectious Diseases (NIAID); HHSH250201700006C from the Health Resources and Services Administration (HRSA); and N00014-20-1-2705 and N00014-20-1-2832 from the Office of Naval Research. We greatly thank Lucia D’Aprano, for her IT assistance in the development of the HLA mutational pipeline. We deeply thank all the reviewers who constructively contributed to improve this work.

Author information

Authors and Affiliations

Authors

Contributions

SP designed the study, collected, analyzed and interpreted the data, developed the pipeline for the variant calling in HLA region, performed the bioinformatic and statistical analyses, and wrote the manuscript. CG performed NGS experiments, clinical and molecular data collection and participated in the analysis interpretation and critical manuscript revision. CH developed NovoHLA and the methodology for the calculation of copy number variation. SH helped in optimizing the bioanalytical HLA workflow and developing the associated bioinformatic pipeline. NN performed WGS experiments, AW, LT, MM helped in sample and data collection; SS and SG provided HLA and SNP array data for the TOAA/CIBMTR IAA cohort and gave helpful intellectual insights. WZ analyzed SNP-array data. VV helped in data collection and interpretation, gave important intellectual inputs, and edited the manuscript; CZ and PZ provided single-cell RNA samples and gave meaningful intellectual inputs for the deployed analytical methodology. TH supervised the genotyping experiments in MLL, helped in data interpretation and edited the manuscript; JPM designed and conceptualized the study, supervised genomic experiments, provided funding and resources, interpreted the data analysis and edited the manuscript.

Corresponding author

Correspondence to Jaroslaw P. Maciejewski.

Ethics declarations

Competing interests

This research was conducted in absence of any commercial or financial relationships that could be construed as a potential conflict of interest. Collaboration between Cleveland Clinic and Novocraft was based on a no-profit academic partnership.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pagliuca, S., Gurnari, C., Hercus, C. et al. Molecular landscape of immune pressure and escape in aplastic anemia. Leukemia 37, 202–211 (2023). https://doi.org/10.1038/s41375-022-01723-w

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41375-022-01723-w

  • Springer Nature Limited

This article is cited by

Navigation